US6518245B1 - Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase - Google Patents

Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase Download PDF

Info

Publication number
US6518245B1
US6518245B1 US09/016,145 US1614598A US6518245B1 US 6518245 B1 US6518245 B1 US 6518245B1 US 1614598 A US1614598 A US 1614598A US 6518245 B1 US6518245 B1 US 6518245B1
Authority
US
United States
Prior art keywords
inhibitor
cam kinase
administration
calmodulin
antiarrhythmic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
US09/016,145
Other languages
English (en)
Inventor
Mark E. Anderson
Andrew P. Braun
Howard Schulman
Ruey J. Sung
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Leland Stanford Junior University
Original Assignee
Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Leland Stanford Junior University filed Critical Leland Stanford Junior University
Priority to US09/016,145 priority Critical patent/US6518245B1/en
Priority to US10/353,759 priority patent/US7251525B2/en
Application granted granted Critical
Publication of US6518245B1 publication Critical patent/US6518245B1/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics

Definitions

  • the present invention relates to methods for the treatment of arrhythmias by inhibition of a multifunctional calcium/calmodulin-dependent protein kinase (CaM kinase), pharmaceutical compositions useful in such treatments, and methods for identifying new agents useful for such treatments.
  • CaM kinase multifunctional calcium/calmodulin-dependent protein kinase
  • Arrhythmias are a leading cause of cardiac-related death in the United States. Prolongation of the cardiac action potential is an important predisposing condition for these arrhythmias. Many antiarrhythmic drugs directly prolong the action potential duration and so may further contribute to these arrhythmias (i.e. the proarrhythmic effects of antiarrhythmic drugs). Despite their cost, implantable cardiac defibrillators (ICDs) have become the treatment of choice for arrhythmias. In order to prevent painful shocks ⁇ 50% of patients with ICDs require additional treatment with antiarrhythmic drugs. Thus there is an important need to develop better antiarrhythmic drug therapies.
  • ICDs implantable cardiac defibrillators
  • EADs Early afterdepolarizations (EADs) are depolarizing oscillations in the action potential (AP) that occur during repolarization.
  • I Ca inward L-type Ca 2+ current
  • I Ca is present at cell membrane potentials (Vm) within the window of I Ca steady state activation and inactivation overlap; such as occur during action potential repolarization.
  • Vm cell membrane potentials
  • Prolongation of action potential repolarization may increase the time that the Vm is in the window current range for I Ca and thus the likelihood of EADs.
  • EADs are important because they are one probable cause of lethal arrhythmias associated with long QT intervals including torsade de pointes.
  • a long QT interval reflects prolonged action potential repolarization in ventricular myocardium and is due to a wide variety of conditions including bradycardia and hypokalemia.
  • One important cause of long QT intervals are antiaffhythmic drugs and the ventricular proarrhythmic effects of many antiarrhythmic agents are due to QT interval prolongation.
  • Intracellular Ca 2+ increases simultaneously with EADs in isolated ventricular myocytes (De Ferrari et al. (1995) Circ 91:2510-2515). Elevation of intracellular Ca 2+ ([Ca 2+ ] i ) has complex effects on I Ca including indirect enhancement through a multifunctional Ca 2+ /calmodulin-dependent protein kinase II pathway (Anderson et al. (1994) Circ Res 75:854-861) and direct inactivation.
  • CaM kinase II The net effect of elevated [Ca 2+ ] i in rabbit ventricular myocytes following flash photolysis of the photolabile Ca 2+ chelator Nitr ⁇ 5 is 40-50% augmentation of peak I Ca that is mediated by a multifunctional Ca 2+ /calmodulin-dependent protein kinase II, hereafter referred to as CaM kinase II.
  • CaM kinase II like other multifunctional Ca 2+ /calmodulin-dependent protein kinases, is an ubiquitous serine-threonine kinase that is activated when Ca 2+ is bound to the Ca 2+ binding protein calmodulin.
  • CaM kinase II activation may be sustained by intersubunit enzyme autophosphorylation that confers Ca 2+ -independent activity, allowing for its activity to persist during the long diastolic intervals associated with QT interval prolongation and torsade de pointes.
  • This Ca 2+ -independent activity is enhanced by long stimulating pulses (De Koninck and Schulman (1998) Science 279:227-230), as occur with prolonged action potential repolarization.
  • EADs caused by I Ca may be enhanced by increased [Ca 2+ ] i through the CaM kinase II pathway.
  • EADs can occur in conditions adverse to CaM kinase activity such as enhanced [Ca 2+ ] i buffering.
  • DADs Delayed afterdepolarizations
  • Intracellular calcium overload is a central feature of many ventricular arrhythmias occurring during ischemia (Lee et al. (1988) Circ 78:1047-1059) including ventricular fibrillation.
  • Inhibition of CaM kinase activity can be used to test for a facilitatory role of CaM kinase in EADs and DADs.
  • Synthetic pseudo-substrate peptide inhibitors of CaM kinase provide a specific approach to CaM kinase inhibition and have been used in a variety of cell types including ventricular myocytes (Braun et al. (1995) J. Physiol 488:37-55).
  • the peptide sequence KKALHRQEAVDCL SEQ ID NO:1
  • SEQ ID NO:1 is a much less efficient inhibitor of both protein kinase A (PKA) and protein kinase C (PKC), with an IC 50 value of at least 500 ⁇ mol/l for each.
  • PKA protein kinase A
  • PKC protein kinase C
  • Myristoylated inhibitory peptides are cell membrane permeant and thus could also be effective when added extracellularly.
  • KN-93 (2-[N-(2-hydroxyethyl)-N-(4-methoxy-benzenesulfonyl)]-amino-N-(4-chlorocinnamyl)-N-methylbenzylamine) is a methoxybenzene sulfonamide derivative that competitively inhibits calmodulin binding to CaM kinase with a reported K i of 0.37 ⁇ mol/l.
  • KN-93 has been shown to inhibit CaM kinase—dependent processes in PC12h cells, fibroblasts, and gastric parietal cells.
  • KN-92 (2-N-(4-methoxybenzenesulfonyl)-amino-N-(4-chlorocinnamyl)-N-methylbenzylamine) is a congener of KN-93 without CaM kinase inhibitory activity and is used as an experimental control.
  • KN-92 Direct I Ca blockade by KN-92 has also not been previously reported, and neither KN-93 nor KN-92 have appreciable effects on other serine threonine kinases such as protein kinase A (PKA) or protein kinase C (PKC).
  • PKA protein kinase A
  • PKC protein kinase C
  • PKA protein kinase A
  • action potential duration is governed by a number of different ionic currents and it is not typically known which current is critical in a given patient.
  • specific antiarrhythmic drugs are not available for modification of many of these ionic currents.
  • Isoproterenol also does not always shorten the action potential, can itself be arrhythmogenic, may cause ischemia, and can only be used in an acute setting.
  • suppression of PKA is accomplished indirectly by stellate gangionectomy or by ⁇ -adrenergic antagonists.
  • ⁇ -adrenergic antagonists are effective in reducing death from arrhythmias, but application is limited by the fact that these agents weaken the force of heart muscle contraction.
  • Ventricular arrhythmias due to ischemia and intracellular calcium overload are generally treated with revascularization (i.e. coronary artery angioplasty or coronary artery bypass surgery), ⁇ -adrenergic antagonists, and class III antiarrhythmic medications (e.g. sotalol and amiodarone), in addition to ICDs.
  • revascularization i.e. coronary artery angioplasty or coronary artery bypass surgery
  • class III antiarrhythmic medications e.g. sotalol and amiodarone
  • ICDs intracellular calcium overload
  • Class III antiarrhythmic agents may be proarrhythmic by causing excessive action potential prolongation or be associated with use—limiting toxicity (e.g. amiodarone).
  • Beta-adrenergic antagonist use is also limited as previously discussed (above).
  • Atrial fibrillation is associated with significant morbidity and mortality from stroke and heart failure. Atrial fibrillation can be caused by DADs. Maintenance of atrial fibrillation is favored by intracellular calcium dependent processes. (Tieleman et al. (1997) Circ 95:1945-1953) Present conventional therapies center around anticoagulation (for prevention of stroke), ventricular rate control, and antiarrhythmic agents. Currently available antiarrhythmic agents only succeed in maintaining sinus rhythm in ⁇ 50% of patients/year. Recent experimental therapies include artificial pacing systems and atrial ICDs.
  • CaM kinase inhibition may be superior to previous antiarrhythmic strategies because CaM kinase has characteristics that may allow it to function as a proarrhythmic positive feedback effector for EADs and DADs, and thus play a more central role in EAD and DAD induction than other effectors, such as PKA.
  • L-type calcium current increases intracellular calcium directly by transmembrane calcium flux and indirectly through calcium-induced release of calcium from the sarcoplasmic reticulum. Increased intracellular calcium results in enhanced CaM kinase activity to further favor EADs during action potential prolongation. In contrast, PKA activity is not enhanced by increased intracellular calcium.
  • CaM kinase inhibition may be highly beneficial as a treatment for arrhythmias related to excessive action potential prolongation and EADs and for arrhythmias related to intracellular calcium overload such as atrial and ventricular fibrillation.
  • L-type Ca 2+ current inhibitors have not been found to be highly effective antiarrhythmic agents for atrial and ventricular fibrillation as well as most types of ventricular tachycardia. Lack of ventricular antiarrhythmic efficacy for I Ca antagonists at clinically tolerated doses may be because the amount of I Ca inhibition is insufficient to prevent or terminate most EADs. Combination of a I Ca antagonist with a CaM kinase inhibitor may be effective, however.
  • the present invention provides methods for treating and preventing arrhythmias in a human subject through the administration of a multifunctional calcium/calmodulin-dependent protein kinase inhibitor to the human in an amount sufficient to suppress early after depolarizations, delayed afterdepolarizations, or intracellular calcium overload.
  • compositions comprising a multifunctional calcium/calmodulin-dependent protein kinase inhibitor and a pharmaceutically acceptable excipient.
  • Another aspect of the invention provides for methods of identifying agents useful in the treatment of arrhythmias and provides feasible strategies for the development of a therapeutic, antiarrhythmic drug which can be applied systemically.
  • the present invention provides for, methods of treating and preventing arrhythmias in which a multifunctional calcium/calmodulin-dependent protein kinase inhibitor is administered in combination with a second antiarrhythmic agent in order to increase safety and efficacy of the treatment.
  • the present invention provides for methods of treating and preventing arrhythmias in which the administration of a multifunctional calcium/calmodulin-dependent protein kinase inhibitoris is combined with treatment of the patient with an antiarrhythmic device.
  • the present invention also provides for the administration of a multifunctional calcium/calmodulin-dependent protein kinase inhibitor in combination with a second antiarrhythmic agent to block the proarrhythmic effects due to action potential prolongation caused by the second antiarrhythmic agent.
  • FIG. 1 (A) Experimental design for early afterdepolarizations (EAD) induced by clofilium in an isolated rabbit heart pretreated with, the inactive analog, KN-92. Data tracings show (from top to bottom) moniophasic action potentials (MAP) and left ventricular (LV) pressure. The horizontal lines to the left of the, LV pressure tracings (top to bottom) indicate 100 mm Hg and 0 mm Hg levels and 850 ms for panels A and B. All experiments with isolated hearts consisted of 3 periods. A fourth period (experimental period 4) was only added in hearts with EADs. Experimental periods are labeled 1-4 (top of page for both panels A and B). Experimental period 1 measurements represent control and follow >10 min of stabilization.
  • MAP moniophasic action potentials
  • LV left ventricular
  • Experimental period 2 measurements are taken 10 min after addition of the inactive KN-93 analog, KN-92 (0.5 ⁇ mole/l).
  • Period 3 measurements follow addition of clofilium (7.5 ⁇ mole/l) and are taken at EAD initiation or from the longest action potential duration recorded over 30 min. Multiple EADs are seen as 2 oscillations during repolarization on the MAP tracings in experimental period 3. Secondary elevations in LV pressure coinciding with EADs are also seen in the LV pressure tracing in experimental period 3.
  • Experimental period 4 measurements follow EAD termination by addition of nifedipine (10 ⁇ mole/l) (shown above) or Cd 2+ (200-500 ⁇ mole/l) (data not shown).
  • Data tracings show MAP with single EADs (top) and LV pressure (bottom).
  • the horizontal lines to the left of the LV pressure tracings indicate 50 mm Hg and 0 mm Hg pressure levels (top to bottom).
  • EAD termination occurs prior to the first discernible decrease (i.e. >10% below baseline) in LV pressure.
  • the vertical arrow marks the first beat of EAD termination.
  • FIG. 2 Left ventricular developed pressure (LVDP, panel A) and interbeat intervals (panel B) in isolated rabbit hearts.
  • LVDP is defined as the peak systolic pressure minus the end diastolic pressure.
  • Numerals 1-4 correspond to the experimental periods illustrated in FIGS. 1A and B.
  • LVDP at experimental period 4 is taken from the first beat after EAD termination as shown in FIG. 1C. A.
  • Hearts pre-treated with KN-93 failed to increase LVDP following clofilium treatment failed to increase LVDP following clofilium treatment (panel A, experimental period 3).
  • FIG. 3 Monophasic action potential durations (MAP) at 50% (APD 50 , circles) and 90% (APD 90 , squares) repolarization to baseline in isolated rabbit hearts.
  • Numerals 1-4 correspond to the experimental periods illustrated in FIGS. 1A and B.
  • Filled symbols are for hearts pretreated with the inactive analog KN-92.
  • Open symbols are for hearts pre-treated with the CaM kinase inhibitor KN-93.
  • the data set is the same as in FIG. 2.
  • *p 0.004 and **p ⁇ 0.001 compared with corresponding APDs in period 1.
  • No significant differences in MAP durations were present between hearts pretreated with the CaM kinase inhibitor KN-93 or the inactive analog KN-92.
  • MAP durations were not significantly different during early afterdepolarizations (EAD) and the first beat after EAD termination by nifedipine (10 ⁇ mole/l) or Cd 2+ (200-500 ⁇ mole/l).
  • FIG. 4 Percent calcium-independent CaM kinase activity in isolated rabbit hearts. Maximal (i.e. Ca 2+ /calmodulin-dependent) and Ca 2+ -independent CaM kinase activities were assayed from left ventricular (LV) tissue homogenates as described in Example 5, Experimental Methods. All hearts were initially isolated under control conditions. A stabilization period of ⁇ 10 min was used for each isolated heart to ensure the absence of EADs, as indicated by monophasic action potentials (MAPs) recorded by a catheter positioned over the LV epicardium.
  • MAPs monophasic action potentials
  • FIG. 5 Effects of KN-93 and KN-92 on CaM kinase activity in vitro. CaM kinase activity in left ventricular tissue homogenate was assayed as described in Example 5, Experimental Methods. The plot shows that increasing concentrations of KN-93, but not the inactive analog KN-92, produce direct inhibition of cardiac CaM kinase activity. The solid line represents a fit to the KN-93 data using the Hill equation. The dashed line shows the expected inhibition of CaM kinase activity after accounting for the concentration of added calmodulin in the assay (150 ⁇ mole/l), given the competitive interaction between KN-93 and calmodulin with CaM kinase.(Sumi et al. (1991) Biochem Biophys Res commun 181: 968-975) The K i value for inhibition by KN-93 calculated from the dashed curve is 2.58 ⁇ mole/l.
  • FIG. 6 KN-93 and KN-92 effects on L-type Ca 2+ current (I Ca ).
  • A Peak steady state current voltage (I-V) relationship for I Ca during control conditions (filled squares) and following addition of the inactive agent KN-92 (0.5 ⁇ mol/l filled circles and 1.0 ⁇ mol/l filled diamonds) to the cell bath.
  • the inset shows superimposed raw currents and the command voltage step (300 ms) from the ventricular myocyte used to construct the I-V relationship.
  • the horizontal bar marks the zero current level.
  • B Tracings are laid out as in A.
  • KN-93 and KN-92 were equipotent peak steady state I Ca inhibitors at the concentration used to suppress early afterdeploarizations in this study, but KN-93 was a more effective I Ca inhibitor at 1.0 ⁇ mol/l.
  • D Time dependence of I Ca recovery from inactivation. Peak I Ca was elicited from a holding potential of ⁇ 80 mV to atest potential of 0 mV for 300 ms (P1). Progressively longer intervals (10-2500 ms) were inserted between P1 and a second pulse (P2). Peak I Ca during P2 was expressed as a fraction of peak I Ca during P1. Data are plotted for control conditions (filled squares) in the presence of 0.5 ⁇ mol/I KN-92 (filled circles) or 0.5 ⁇ mol/l KN-93 (open circles).
  • FIG. 7 CaM kinase inhibition suppresses pacing induced inward current.
  • a prolonged action potential was digitized and used as a voltage clamp command waveform for current tracings below.
  • Command membrane potential is on the ordinate and time (for panels A-C) is on the abscissa
  • B Inward currents developed after completion of the command wave form in 5/6 cells dialyzed with a control peptide (SEQ ID NO:2, 20 ⁇ mole/l) devoid of CaM kinase inhibitory activity. Cell membrane currents are referenced on the ordinate for both panels B and C.
  • the present invention also provides for the prevention of the occurrence of arrhythmias through the administration of a multifunctional calcium/calmodulin-dependent protein kinase inhibitor to the patient patient in an amount sufficient to suppress early afterdepolarizations, delayed afterdepolarizations, or intracellular calcium overload.
  • CaM kinase is used herein to refer to any member of the multifunctional calcium/calmodulin-dependent protein kinase family in general, whereas the term “CaM kinase II” is used herein only to refer to the multifunctional calcium/calmodulin-dependent protein kinase II (and its isoforms).
  • an “inhibitor” of CaM kinase is a compound capable of decreasing the activity of the enzyme (generally via noncovalent binding of the inhibitor to the enzyme).
  • the inhibitor may provide reversible inhibition. Reversible inhibition can consist of competitive inhibition, noncompetitive inhibition, uncompetitive inhibition, or mixed inhibition.
  • An “inhibitor” of CaM kinase can also be an irreversible inhibitor or it can be a suicide inhibitor.
  • an inhibitor of a multifunctional calcium/calmodulin-dependent protein kinase II is used in the treatment and prevention of arrhythmias.
  • the use of compounds which inhibit other members of the family of multifunctional calcium/calmodulin-dependent protein kinases is also provided for by the present invention.
  • the family of multifunctional calcium/calmodulin-dependent protein kinases includes CaM kinase I, CaM kinase II, and CaM kinase IV. Each of these members also additionally exists in a multitude of isoforms.
  • An inhibitor of one of the multifunctional calcium/calmodulin-dependent protein kinases is generally understood by one skilled in the art to be likely to inhibit the other members of the family as well, due to structural conservation amongst the types (Enslen et al. (1994) J. Biol. Chem . 269:15520-15527).
  • isoform-specific inhibitors could be used as agents with enhanced specificity in the prevention and treatment of arrhythmias (Braun et al. (1995) J. Physiol 488:37-55).
  • organ-specific CaM kinase inhibtors could be used. Cardiac specificity of a multifunctional calcium/calmodulin-dependent protein kinase may be achieved by combining a CaM kinase inhibitor with cardiac cell specific epitope recognition sequences. The use of organ or isoform specific inhibitors may help ensure efficacy and the safety of systemic delivery of the drug.
  • CaM kinase inhibitors such as KN-93 and any of the inhibitory peptides such as KKALHRQEAVDCL (SEQ ID NO:1) can be used in the methods of treating arrhythmias described herein. Both inhibitors act competitively, but at distinct sites on the kinase. KN-93 binds and prevent activation by calmodulin, whereas SEQ ID NO:1 binds to the catalytic site, preventing interaction with substrate molecules.
  • Other general, organ-specific, or isoform-specific CaM kinase inhibitors may also be used in the treatment of arrhythmia as provided for by the present invention.
  • the inhibitor is delivered systemically.
  • the CaM kinase inhibitor is delivered intravenously.
  • Other possible methods of delivery include, but are not limited to oral delivery, bolus injection or continuous parenteral pump infusion, cutaneous patch or cutaneous iontophoretic delivery, and intracoronary or intrapericardial delivery, or gene transfer therapy, as recently described by Griffith et al. (Griffith et al. (1993) Neuron 10:501-509).
  • an “arrhythmia” is a disturbance in the heart's natural rhythm.
  • the arrhythmia may be any atrial or ventricular arrhythmia associated with a prolonged action potential, early afterdepolarizations, delayed afterdepolarizations, or intracellular calcium overload.
  • Cardiac conditions which involve such arrhythmias include, but are not limited to, long QT syndrome, cardiomyopathy, and the proarrhythmic effects of certain medications including antiarrhythmic drugs and ischemia.
  • Other structural abnormalities of the heart, abnormalities of the heart's electrical system, and diseases may also cause arrhythmias treatable by the present invention.
  • One particular clinical indication for intervention is the presence of prolonged QT intervals (>480 ms). The arrhythmia to be treated may be life-threatening.
  • a preferred embodiment of the present invention is the treatment of ventricular tachycardia, an arrhythmia associated with early afterdepolarizations. Treatment of atrial and ventricular fibrillation, arrhythmias associated with DADs and intracellular overload are also provided for by the invention.
  • a “sufficient amount” or an “effective amount” of a multifunctional calcium/calmodulin-dependent protein kinase inhibitor according to this invention is an amount sufficient to suppress early afterdepolarizations (EADs), delayed afterdepolarizations (DADs), or intracellular calcium overload.
  • EADs early afterdepolarizations
  • DADs delayed afterdepolarizations
  • intracellular calcium overload early afterdepolarizations
  • Early afterdepolarizations are depolarizing oscillations in the action potential and their likelihood of occurrence is increased by the prolongation of action potential repolarization. They are also a probable cause of some lethal arrhythmias. Delayed afterdepolarizations are a marker of intracellular calcium overload and also the cause of some lethal arrhythmias.
  • an effective amount of inhibitor will vary with the activity of the inhibitor.
  • from about 0.05 to about 5.0 mg of inhibitor per kilogram of subject's body weight is administered.
  • from about 0.3 to about 3.0 mg per kilogram is administered.
  • this dosage could be repeated every thirty to sixty minutes until suppression of early afterdepolarizations is achieved, unless limited by the development of hypotension due to the Ca channel blocking effect.
  • a myristoylated CaM kinase inhibitory peptide is administered at a dose of from about 2 to about 20 ⁇ mole per kilogram of body weight.
  • the administration of a CaM kinase inhbitor may also be combined with administration of a second antiarrhythmic agent to the patient.
  • This combination may provide benefits both in terms of efficacy and safety.
  • the CaM kinase inhibitor is administered to the patient in conjunction with a second antiarrhythmic agent that is known to have a proarrhythmic effect.
  • Existing antiarrhythmic agents which may be delivered to a patient in combination with the CaM kinase inhibitor include K-channel blockers, such as the class IA or III antiarrhythmic drugs.
  • the class IA agents procainamide (1.0 gram i.v.
  • the class III agents dofetilide (8 ⁇ g/Kg i.v.) or d-sotalol (5.7 mg/Kg/24 hours p.o.) would be administered with a CaM kinase inhibitor.
  • the administration of a CaM kinase inhibitor may also be combined with administration of a ⁇ -adrenergic antagonist to increase efficacy of antiarrhythmic therapy.
  • the ⁇ -adrenergic antagonist atenolol (50-100 mg/day p.o./i.v.), propranalol (60 mg/6 hours p.o. or 30 mg/6 hours i.v.), or metoprolol (50-100 mg/6 hours p.o./i.v.) would be administered with a CaM kinase inhibitor.
  • the administration of a CaM kinase inhibitor may also be combined with administration of a calcium channel blocker to increase efficacy of antiarrhythmic therapy.
  • the calcium channel antagonist nifedipine (10-30 mg/6 hours p.o.), verapamil (80-120 mg/6 hours p.o. or 0.075-0.15 mg/Kg i.v.), diltiazem (30-90 mg/6 hours p.o. or 0.075-0.15 mg/Kg i.v.), mibefradil (50-100 mg/day p.o.) would be administered with a CaM kinase inhibitor.
  • the administration of a CaM kinase inhibitor may be combined with a treatment that utilizes an antiarrhythmic device. This combination is useful in treating and preventing arrhythmias.
  • the antiarrhythmic device is an implantable cardiac defibrillator (also referred to herein as an implantable cardioverter defibrillator).
  • the administration of a CaM kinase inhibitor may be combined with an implantable atrial cardiodefibrillator to treat atrial fibrillation.
  • the administration of a CaM kinase inhibitor may be combined with an implantable ventricular cardiodefibrillator to treat ventricular fibrillation.
  • the administration of a CaM kinase inhibitor may be combined with a permanent pacing system to prevent atrial fibrillation.
  • the present invention provides for a pharmaceutical composition of a calcium/calmodulin-dependent protein kinase inhibitor and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition of a calcium/calmodulin-dependent protein kinase inhibitor and a pharmaceutically acceptable excipient.
  • an inhibitor of CaM kinase II is used.
  • a “pharmaceutically acceptable excipient” or “carrier” is a therapeutically inert substance which serves as a diluent or delivery vehicle for the inhibitor drug. Selection of suitable carriers for the pharmaceutical composition is readily achievable by one skilled in the art and would in part be dictated by the specific CaM kinase inhibitor chosen. Typical pharmaceutical compositions useful for CaM kinase II inhibitors are disclosed in Remington, The Science and Practice of Pharmacy , 19th ed., 1995, Mack Publishing Co., Easton, Pa., the disclosure of which is incorporated by reference herein.
  • Possible pharmaceutical excipients include polymers, resins, plasticizers, fillers, binders, lubricants, glidants, disintegrants, solvents, co-solvents, buffer systems, surfactants, preservatives, sweetening agents, flavoring agents, pharmaceutical grade dyes or pigments, and viscosity agents.
  • a method for identifying agents useful in the treatment or prevention of arrhythmias is also provided by the present invention. Screening for novel antiarrhythmia agents may be conducted by determining if the agent inhibits CaM kinase II activity.
  • One test for CaM kinase II inhibition is described herein; however, alternative methodologies for identifying inhibitors via exposure to CaM kinase II are within the purview of this invention.
  • the test for CaM kinase II inhibition may be done in vivo, but is preferably done in vitro. The ability of an assayed compound to inhibit the activity of CaM kinase II to an appreciable degree indicates that the compound will have antiarrhythmic properties.
  • the invention also provides for methods of suppressing early and delayed afterdepolarizations and intracellular calcium overload in a mammal. This suppression is achieved by the administration of an inhibitor of a multifunctional calcium/calmodulin-dependent protein kinase to the subject.
  • the inhibitor is a CaM kinase II inhibitor.
  • Another aspect of the invention provides for methods of treating arrhythmia in a human by decreasing the activity of a CaM kinase in the human.
  • This decrease in activity may be achieved through a number of ways, each readily discernible by one skilled in the art.
  • the activity of CaM kinase is decreased by the administration of a CaM kinase inhibitor to the subject.
  • the activity of a CaM kinase may be achieved through other means such as by decreasing the level of expression of a CaM kinase in a human by an antisense strategy or similar method.
  • the present invention in all aspects, is enabled by our discovery that EADs and DADs are due to I Ca and/or intracellular calcium overload and are facilitated by a multifunctional calcium/calmodulin-dependent protein kinase.
  • EADs were monitored with monophasic action potential (MAP) catheters. Reliable EAD induction was achieved using hypokalemia, bradycardia, and the Vaughn-Williams class III antiarrhythmic agent clofilium in isolated perfused rabbit hearts.
  • I Ca I Ca antagonists nifedipine and Cd 2+
  • EAD termination by I Ca antagonists appeared to be a direct effect of I Ca blockade, and not a secondary effect of [Ca 2+ ] i depletion, because of its 1) rapid onset (FIG. 1) and 2) occurance before significant loss of LVDP (FIG. 2 ).
  • the lack of MAP shortening at EAD termination is further evidence that EAD termination is a direct effect of I Ca blockade and not due to a secondary effect on MAP duration (FIG. 3 ).
  • the clofilium-induced increase in action potential duration is expected to increase Ca 2+ entry via I Ca .
  • CaM kinase interacts with other [Ca 2+ ] i dependent processes in ventricular myocytes. CaM kinase both facilitates Ca 2+ uptake by the sarcoplasmic reticulum and release by the ryanodine receptor.
  • the anticipated net effect of CaM kinase activation in ventricular myocytes is increased LVDP.
  • Clofilium did increase LVDP in KN-92 but not KN-93 pretreated hearts (FIG. 2 ).
  • CaM kinase II undergoes intersubunit autophosphorylation which results in Ca 2+ -independent CaM kinase II activity.
  • Measurement of Ca 2+ -independent activity by CaM kinase thus, provides an important measure of CaM kinase activation.
  • CaM kinase inhibitor KN-93 Example 4, FIG. 4
  • FIG. 1 A The experimental design for the isolated heart experiments is shown in FIG. 1 A. All clofilium-induced EADs were terminated by the I Ca antagonists Cd 2+ (200-500 ⁇ mol/l) or nifedipine (10 ⁇ mol/l). EAD termination occurred in ⁇ 10 sec from a discernable decrease (>10% from preceeding baseline) in LVDP after addition of I Ca antagonist to the perfusate. EAD termination occurred before LVDP decreased significantly (FIG. 2A) and without a change in the interbeat interval (FIG. 2 B). Action potential duration was not shortened upon the first beat of EAD termination.
  • EAD termination by I Ca antagonists was unlikely due to a secondary effect on [Ca 2+ ] i because LVDP was not significantly depressed at initial termination.
  • EAD termination by I Ca antagonists was not due to secondary effect on MAP duration or heart rate.
  • New Zealand White rabbits of either gender were treated with a heparin bolus (150 U/kg IV) and killed by pentobarbital (50 mg/kg IV) overdose.
  • Hearts were rapidly excised and placed in ice-cold Tyrode's solution for dissection of extracardiac tissue.
  • Tyrode's solution was composed of (mmol/l) NaCl (130.0), NaHCO 3 (20.0), glucose (5.6), KCl (3.0), CaCl 2 (2.0), NaH 2 PO 4 (1.8), MgCl 2 4H 2 O (0.7).
  • the aorta was cannulated, and perfuse retrograde with Tyrode's solution.
  • the perfusate (200 ml) was recirculated through a warming bath and filling pressure was adjusted to 20 cm of H 2 O. Recirculation time was ⁇ 1 min, based on the time to decrease in left ventricular (LV) pressure following addition of I Ca blockers to the recirculating perfusate. Temperature was monitored using a thermistor probe positioned in the right ventricle (RV) and maintained at 33 ⁇ 1° C. by a closed loop feed back system. The perfusate was vigorously bubbled with 95% O 2 , 5% CO 2 and pH was monitored throughout the experiments and adjusted to 7.4 with 1N NaOH or HCl, as appropriate. Both atria were removed and drains were placed in the RV and LV apices.
  • An electrocardiogram (ecg) lead was sutured to the LV apex and the proximal pulmonary artery.
  • Epicardial pacing wires were inserted into the RV free wall. Bradycardia was produced by crushing the AV node with a forceps.
  • Epicardial pacing maintained a minimum cycle length of 1500 ms (heart rate of 40 beats/min). Pacing output was adjusted to twice diastolic threshold.
  • LV pressure was continuously monitored with a solid state transducer (Camino Laboratories) placed in a saline filled latex balloon.
  • LVDP peak systolic pressure—peak diastolic pressure
  • a single MAP electrode (EP technologies Inc., Sunnyvale, Calif.) was positioned over the LV epicardium with a macro-manipulator. The heart was positioned against an adjustable stop opposite the epicardial MAP catheter to minimize heart motion.
  • the LV balloon was omitted and paired MAP catheters were positioned opposite one another on the LV epicardial and endocardial free wall.
  • MAP recordings were concordant for the presence and absence of EADs in all but one instance, suggesting that epicardial MAP recording is a valid method for detecting EADs from LV epicardium and endocardium in this isolated heart model.
  • MAP signals were amplified from (0 to 100 Hz) with a direct current coupled preamplifier (EP technologies Inc., Sunnyvale, Calif.) and recorded directly onto a chart recorder (Gould 2600S) at 100 mm/sec. Data from some experiments were digitized (Neuro-corder) and stored on a videotape recorder. Acceptable MAP catheter position was confirmed by a stable action potential configuration free of EADs or delayed after depolarizations (DADs) during a >10 minute control period. The MAP amplitude was defined as the difference between phase 2 and phase 4. MAP amplitude at the start of the experiments was 10.4 ( ⁇ 1.26) mV.
  • the MAP duration was measured from the onset of phase 0 to the point of 50% (APD 50 ) and 90% (APD 90 ) repolarization.
  • EADs were included in the APD measurements. All MAP durations are reported as the mean of 3 contiguous beats, except following EAD termination where the first beat after EAD termination by Cd 2+ or nifedipine was used.
  • Beat to beat intervals were measured from the onset of phase 0 using contiguous MAPs. Beat to beat intervals during irregular rhythms are reported as the longest intervals present during the measurement period.
  • EADs were defined as discrete oscillations in MAP repolarization with the slope of the tangent to the onset of the oscillation >0.0 (FIG. 1 ). EADs had to be present on most (>90%) beats over a 1 min period or in a stable bigeminal alternating pattern to be classified as inducible. Once these criteria were met EADs continued for >2 min and always required I Ca blockade for termination. Multiple EADs were defined as >1 EAD ocurring per beat (FIG. 1 ). The CaM kinase inhibition experiments consisted of 4 periods (FIG. 1 A). The first period (>10 min) was the control and established stable baseline values for MAPs, LVDP, heart rate, and rhythm.
  • the second period followed addition of KN-92 or KN-93 to the perfusate and lasted 10 min.
  • the third period followed addition of clofilium to the perfusate and lasted for 30 min or until EADs occurred. If no EADs occurred then the longest MAP durations were used. MAP and LVDP measurements were made at 10 min intervals and at the time of EAD occurrence during the third period.
  • the fourth period only consisted of experiments where prior EADs occurred, and measurements followed addition of nifedipine (10 ⁇ mol/l) or Cd 2+ (200-500 ⁇ mol/l) to the perfusate.
  • EAD induction by clofilium was not different in hearts pretreated with KN-92 (EADs in 10/11 hearts) compared to hearts without any type of pretreatment (EADs in 8/8 hearts), suggesting that the inactive analog KN-92 had no effect on EAD inducibility by clofilium (data not shown).
  • Left ventricular developed pressure increased significantly, compared with control, following addition of clofilium to the bath in hearts pretreated with the inactive. analog KN-92 (FIG. 2 A).
  • Left ventricular developed pressure increases occurred in step with MAP duration prolongation (FIG. 3) and EADs, but without a change in heart rate (FIG. 2 B).
  • KN-93 pretreatment completely prevented the increase in LVDP by clofilium (FIG. 2A) without reducing MAP duration prolongation (FIG. 3) or changing heart rate (FIG. 2 B).
  • Increased LVDP during MAP prolongation and EADs is likely due to CaM kinase—mediated I Ca augmentation.
  • Ca 2+ -independent CaM kinase activity is a marker for CaM kinase activation.
  • EADs appear due to I Ca in this isolated heart model, we hypothesized that EADs are associated with increased Ca 2+ -independent CaM kinase activity.
  • Ca 2+ -independent CaM kinase activity increased significantly in hearts demonstrating EADs after clofilium compared to hearts without clofilium treatment or EADs (FIG. 4 ).
  • Left ventricular homogenate was prepared as follows: after removal of the heart, it was placed in nominally Ca 2+ free ice-cold HEPES-buffered Tyrode's solution. One to two grams of the left ventricular free wall was cut away, coarsely minced, and suspended in 5-10 ml of cold homogenization buffer (mmol/l: 20 PIPES, 1 EDTA, 1 EGTA, 2 DTT, 10 sodium pyrophosphate, and 50-100 ⁇ g/ml leupeptin) at pH 7.0. Tissue suspension was homogenized at 4° C. using 3-10 sec bursts of a Polytron with 30 sec pauses in between bursts.
  • cold homogenization buffer mmol/l: 20 PIPES, 1 EDTA, 1 EGTA, 2 DTT, 10 sodium pyrophosphate, and 50-100 ⁇ g/ml leupeptin
  • the homogenate was then centrifuged at ⁇ 10,000 ⁇ g for 20 min at 4° C. using a JA-20 rotor. The supernate was removed and used directly in the assay. Protein concentration was measured by the method of Bradford, using BSA as the standard.
  • Assay of CaM kinase activity was performed essentially as described by Waldmann et al. with a few modifications. The assay was performed in triplicate in a final volume of 50 ⁇ l containing 50 mmol/l PIPES, pH 7.0, 10 mmol MgCl2, 0.1 mg/ml BSA, 10 ⁇ mol/l autocamtide 3 (synthetic kinase substrate), 150 ⁇ mol/l calmodulin, 1 mmol/l CaCl 2 or 1 mmol/l EGTA, and increasing concentrations (0-100 ⁇ mol/l) of KN-93 or KN-92.
  • tissue homogenate 30-50 ⁇ g of tissue homogenate were added per assay tube, and samples were then pre-incubated at 30° C. for 1 min.
  • the kinase reaction was then started by addition of 50 ⁇ mol/l (final) ⁇ 32 P-ATP ( ⁇ 400 cpm/pmol), and incubation was carried out for an additional 1 to 2 minutes.
  • the reaction was terminated by addition of 10 ⁇ l cold trichloroacetic acid (30% w/v), and samples were then placed on ice for ⁇ 2 min. Calcium/calmodulin-dependent phosphorylation of the autocarntide 3 substrate was then quantitated as described.
  • KN-92 and KN-93 are Equipotent Direct Blockers of L-Type Ca 2+ Current
  • KN-92 is a valid control for the direct I Ca blocking effects of KN-93 in the isolated rabbit ventricular myocytes because these agents have equivalent I Ca antagonist effects at the concentration (0.5 ⁇ mole/l) used to suppress EADs (FIG. 6 C).
  • Single ventricular myocytes were prepared as previously described in Anderson et al. (1994) Circ Res , 75:854-861.
  • the standard Ca 2+ containing solution for myocyte preparation was composed of(mmol/l) NaCl 137.0, HEPES (free acid) 10.0, NaH 2 PO 4 0.33, glucose 10.0, KCl 5.4, CaCl 2 1.8, and MgCl 2 2.0.
  • the nominally Ca 2+ free solution was identical, except CaCl 2 was omitted.
  • the low Ca 2+ solution contained 0.2 mmol/l CaCl 2 .
  • the collagenase solutions were prepared with 1% BSA (wt/vol) and ⁇ 60 U/ml collagenase (Worthington Biochemicals) and ⁇ 0.1 U/ml type XIV protease (Sigma Chemical Co).
  • the myocyte bath solution contained (mmol/l) NaCl 137.0, CsCl 20.0, glucose 10.0, HEPES 10.0, KCl 5.4, CaCl 2 1.8, MgCl 2 0.5, and tetrodotoxin 0.03; pH was adjusted-to 7.4 with 10N NaOH.
  • the intracellular pipette solution contained (mmol/l) CsCl 130.0, 4Cs BAPTA 10.0 (Molecular Probes), phosphocreatine 5.0, NaGTP 1.0, and MgATP1.0; pH was adjusted to 7.2 with 10N CsOH.
  • New Zealand White rabbits (2 to 3 kg body weight) were killed by pentobarbital (50 mg/kg IV) overdose.
  • Hearts were rapidly excised and placed in ice cold nominally Ca 2+ free HEPES—buffered myocyte bath solution.
  • the aorta was cannulated, and the heart was perfused in a retrograde fashion with a nominally Ca 2+ free perfusate for 15 minutes at 37° C. This was followed by a 15-min perfusion with collagenase—containing nominally Ca 2+ free solution.
  • Final perfusion was with collagenase—containing low Ca 2+ (0.2 mmol/L) solution.
  • the LV and septum were cut away, coarsely minced, and placed in a beaker containing low Ca 2+ solution with 1% (wt/vol) bovine serum albumin (BSA) at 37° C.
  • BSA bovine serum albumin
  • Myocytes were dispersed by gentle agitation, collected in serial aliquots, and then maintained in standard saline solution containing 1.8 mmol/L Ca 2+ . All solutions were vigorously oxygenated.
  • Isolated quiescent ventricular myocytes were studied with patch-clamp methodology in the whole cell recording configuration by using an Axopatch 1B amplifier (Axon Instruments). Micropipettes were pulled from glass capillary tubing (VWR) and heat-polished to a tip resistance of 1-3 M ⁇ when filled with the intracellular solution.
  • VWR glass capillary tubing
  • the cell membrane was commanded from ⁇ 40 mV to +60 mV for 200 ms in 10 mV steps from a holding potential of ⁇ 40 mV at a stimulation of 1.0 Hz. This protocol was repeated with 60 sec pauses between series of voltage clamp steps, and transmembrane current was sampled at 2 kHz. Voltage clamp protocols and data acquisition were performed using PCLAMP software (version 6.0, Axon Instruments) on a microcomputer (Gateway 2000) with a A/D, D/A convertor (Digidata 1200, Axon Instruments). All experiments were performed at room temperature (20° C. to 23° C.) on the stage of a Diaphot inverted microscope (Nikon corp).
  • Action potentials were induced in isolated rabbit ventricular myocytes studied in current clamp with low (1.0 mM) extracellular potassium to favor EAD induction.
  • EADs were induced in the majority of control cells dialyzed with an inactive peptide (11/15) but not in cells dialyzed with the CaM kinase inhibitory peptide (4/15).
  • EADs seen in cells treated with the control peptide tended to be complex and repetitive (i.e. multiple), but only single EADs were present in inhibitory peptide treated cells.
  • Isolated heart cells (Experimental Methods, Example 6) were stimulated (0.5 Hz) in voltage clamp using a prolonged action potential wave form (FIG. 7 A). Rapid pacing favors intracellular calcium overload. Inward currents that develop in response to rapid pacing are a marker for intracellular calcium overload and they are the basis for arrhythmogenic delayed afterdepolarizations (DADs). The majority of cells (5/6) treated with an inactive control peptide (SEQ ID NO:2) developed inward currents (FIG. 7 B). Treatment of a second group with the active CaM kinase inhibitory peptide (SEQ ID NO:1) completely prevented the inward currents (inward currents in 0/5 cells) (FIG. 7 C).
  • DADs arrhythmogenic delayed afterdepolarizations
  • Action potential recording was performed in the following bath solution (mmol/L): NaCl 140.0, Glucose 10.0, HEPES 5.0, KCl 5.4, CaCl 2 2.5, MgCl 2 1.0; pH was adjusted to 7.4 with 10 N NaOH.
  • the intracellular-pipette solution for action potential recording contained (mmol/L): K aspartate 120.0, HEPES 10.0, EGTA 10.0, Na 2 ATP 5.0, MgCl 2 4.0, CaCl 2 3.0; pH was adjusted to 7.2 with 1N KOH. Unless otherwise noted, all chemicals were from Sigma.
  • Cells were stimulated at 0.1 Hz in whole cell configuration in current clamp mode with 0.1 to 1.0 nA pulses of depolarizing current (1.25 ⁇ threshold) for 2-3 ms at room temperature (20-23° C.).
  • Action potentials were low pass filtered at 2 KHz and sampled at 2.5 KHz with a 12-bit analog to digital converter (Digidata 1200 B, Axon Instruments).
  • a long action potential waveform was digitized and stored for application as a voltage command using pClamp 6.03.
  • Isolated cells were studied with the action potential command wave form using voltage clamp methodology (Experimental Methods, Example 6). Cells were stimulated at 0.5 Hz and 32° C. (heated stage from Warner Instruments) for up to 100 beats. Recording solutions were identical to those for action potential recording except that the calcium buffer EGTA was omitted from the intracellular solution to favor intracellular calcium overload. The presence or absence of the post-repolarization inward current was noted.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Medicines Containing Plant Substances (AREA)
US09/016,145 1997-01-31 1998-01-30 Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase Expired - Fee Related US6518245B1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US09/016,145 US6518245B1 (en) 1997-01-31 1998-01-30 Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase
US10/353,759 US7251525B2 (en) 1997-01-31 2003-01-28 Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US3683397P 1997-01-31 1997-01-31
US09/016,145 US6518245B1 (en) 1997-01-31 1998-01-30 Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/353,759 Division US7251525B2 (en) 1997-01-31 2003-01-28 Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase

Publications (1)

Publication Number Publication Date
US6518245B1 true US6518245B1 (en) 2003-02-11

Family

ID=21890905

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/016,145 Expired - Fee Related US6518245B1 (en) 1997-01-31 1998-01-30 Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase
US10/353,759 Expired - Fee Related US7251525B2 (en) 1997-01-31 2003-01-28 Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/353,759 Expired - Fee Related US7251525B2 (en) 1997-01-31 2003-01-28 Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase

Country Status (8)

Country Link
US (2) US6518245B1 (es)
EP (1) EP0966274B1 (es)
JP (1) JP2001509808A (es)
AT (1) ATE218854T1 (es)
AU (1) AU6257798A (es)
DE (1) DE69805973T2 (es)
ES (1) ES2178157T3 (es)
WO (1) WO1998033491A2 (es)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030018362A1 (en) * 2001-06-15 2003-01-23 Chris Fellows Ablation stent for treating atrial fibrillation
US20030060854A1 (en) * 2001-09-25 2003-03-27 Qingsheng Zhu Evoked response sensing for ischemia detection
US20030069606A1 (en) * 2001-06-15 2003-04-10 Girouard Steven D. Pulmonary vein stent for treating atrial fibrillation
US20030153951A1 (en) * 2002-02-08 2003-08-14 Ideker Raymond E. Methods and devices for treating arrhythmias using defibrillation shocks
US20030216349A1 (en) * 2002-04-18 2003-11-20 Luiz Belardinelli Method for treating arrhythmias
US20040073262A1 (en) * 2000-11-02 2004-04-15 Cardiac Pacemakers, Inc. Technique for discriminating between coordinated and uncoordinated cardiac rhythms
US20040093034A1 (en) * 2002-11-12 2004-05-13 Girouard Steven D. Implantable device for delivering cardiac drug therapy
US20040138648A1 (en) * 2000-12-18 2004-07-15 Cardiac Pacemakers, Inc. Drug delivery system for implantable medical device
US20040158289A1 (en) * 2002-11-30 2004-08-12 Girouard Steven D. Method and apparatus for cell and electrical therapy of living tissue
US20040158290A1 (en) * 2002-11-30 2004-08-12 Girouard Steven D. Method and apparatus for cell and electrical therapy of living tissue
US20040220203A1 (en) * 2003-02-10 2004-11-04 Zaijie Wang Method and composition for potentiating an opiate analgesic
US20040266675A1 (en) * 2001-10-01 2004-12-30 Mark Anderson Use of calmodulin kinase II inhibitors to treat myocardial dysfunction in structural heart disease
US20050043675A1 (en) * 2003-08-21 2005-02-24 Pastore Joseph M. Method and apparatus for modulating cellular metabolism during post-ischemia or heart failure
US20050137626A1 (en) * 2003-12-19 2005-06-23 Pastore Joseph M. Drug delivery system and method employing external drug delivery device in conjunction with computer network
US20050192637A1 (en) * 2004-02-27 2005-09-01 Girouard Steven D. Method and apparatus for device controlled gene expression
US20050245972A1 (en) * 2004-04-29 2005-11-03 Medtronic, Inc. Apparatus and methods for treating arrhythmia and lowering defibrillation threshold
WO2006007444A2 (en) * 2004-06-16 2006-01-19 The Trustees Of Columbia University In The City Of New York CaMKII/CALCIUM CHANNEL BINDING-RELATED COMPOSITIONS AND METHODS
US20060036126A1 (en) * 2004-08-16 2006-02-16 Jeffrey Ross Method and apparatus for modulating cellular growth and regeneration using ventricular assist device
US20060057639A1 (en) * 2004-06-16 2006-03-16 Pitt Geoffrey S CaMKII/calcium channel phosphorylation-related compositions and methods
US20060136028A1 (en) * 2004-12-20 2006-06-22 Jeffrey Ross Epicardial patch including isolated extracellular matrix with pacing electrodes
US20070093875A1 (en) * 2005-10-24 2007-04-26 Cardiac Pacemakers, Inc. Implantable and rechargeable neural stimulator
US20090149900A1 (en) * 2005-04-11 2009-06-11 Julia Moffitt Transvascular neural stimulation device
US20090259268A1 (en) * 2004-08-20 2009-10-15 Gregory Waimong Chan Method and apparatus for delivering combined electrical and drug therapies
US20100016927A1 (en) * 2005-05-16 2010-01-21 Anthony Caparso Transvascular reshaping lead system
US20100113563A1 (en) * 2004-01-30 2010-05-06 Zaijie Wang Method for Treating Pain with a Calmodulin Inhibitor
US7764995B2 (en) 2004-06-07 2010-07-27 Cardiac Pacemakers, Inc. Method and apparatus to modulate cellular regeneration post myocardial infarct
US7981065B2 (en) 2004-12-20 2011-07-19 Cardiac Pacemakers, Inc. Lead electrode incorporating extracellular matrix
US9951061B2 (en) 2013-03-06 2018-04-24 Allosteros Therapeutics, Inc. CaMKII inhibitors and uses thereof
US10759792B2 (en) 2014-09-05 2020-09-01 The Johns Hopkins University CaMKII inhibitors and uses thereof

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1311537A2 (en) * 2000-08-25 2003-05-21 MERCK PATENT GmbH Identification of a cam-kinase ii inhibitor
DE10126405A1 (de) * 2001-05-22 2003-02-27 Trommsdorff Gmbh & Co Physiologisch verträgliche Kalium- und Magnesiumsalze enthaltendes pharmazeutisches Präparat sowie dessen Verwendung zur Prophylaxe und/oder Therapie von QT-Zeitverlängerungen
US8841423B2 (en) 2008-04-28 2014-09-23 University Of Iowa Research Foundation Monoclonal antibodies specific for oxidized calcium/calmodulin dependent protein kinase II
WO2012145075A1 (en) 2011-04-22 2012-10-26 Topera, Inc. Flexible electrode assembly for insertion into body lumen or organ

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4860749A (en) * 1988-01-06 1989-08-29 Wayne State University Tachycardia detection for automatic implantable cardioverter/defibrillator with atrial and ventricular sensing capability
US4880005A (en) * 1985-08-12 1989-11-14 Intermedics, Inc. Pacemaker for detecting and terminating a tachycardia
EP0368233A2 (en) 1988-11-07 1990-05-16 Giocondo Baggio Machine for perching leather
US5089526A (en) * 1983-05-23 1992-02-18 Bristol-Myers Company Antiarrhythmic class III process
US5597848A (en) * 1992-10-01 1997-01-28 Hokuriku Seiyaku Co., Ltd. Benzenesulfonamide derivatives and use thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5089526A (en) * 1983-05-23 1992-02-18 Bristol-Myers Company Antiarrhythmic class III process
US4880005A (en) * 1985-08-12 1989-11-14 Intermedics, Inc. Pacemaker for detecting and terminating a tachycardia
US4880005B1 (en) * 1985-08-12 1996-10-29 Intermedics Inc Pacemaker for detecting and terminating a tachycardia
US4860749A (en) * 1988-01-06 1989-08-29 Wayne State University Tachycardia detection for automatic implantable cardioverter/defibrillator with atrial and ventricular sensing capability
EP0368233A2 (en) 1988-11-07 1990-05-16 Giocondo Baggio Machine for perching leather
US5597848A (en) * 1992-10-01 1997-01-28 Hokuriku Seiyaku Co., Ltd. Benzenesulfonamide derivatives and use thereof

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
Anderson et al., "A Role for L-Type Calcium Current, Calmodulin, and Multifunctional Calcium/Calmodulin-Dependent Protein Kinase in Early After Depolarizations," NASPE Abstracts & Program, 19:685 (1996).
Anderson et al., "Early Afterdepolarizations Due to L-Type Ca2+ Current are Suppressed by Inhibition of the Multifunctional Ca2+/Calmodulin-Dependent Protein Kinase in Rabbit Myocardium," American Heart Association 69th Scientific Sessions, Nov., 1996.
Anderson et al., "Multifunctional Ca2+/Calmodulin-Dependent Protein Kinase Mediates Ca2+/Induced Enhancement of the L-type Ca2+Current in Rabbit Ventricular Myocytes," Circ. Res., 75:854-861 (1994).
Berkow et al. "Merck Manual of Diagnosis and Therapy", Merck Sharp & Dohme Research Laboratories, pp. 475-477, 1992.* *
Berkow et al. Merck Manual of Diagnosis and Tehrapy, Merck Sharp & Dhome Research Laboratories pp. 460-475 (1992).* *
Berkow et al. Merck Manual of Diagnosis and Therapy, Merck Sharp & Dohme Research Laboratories, pp. 479-480, 1992.* *
Berkow et al., "Merck Manual of Diagnosis and Therapy," Merck Sharp & Dohme Research Laboratories, 439-442 (1987).
Braun et al., "A Non-Selective Cation Current Activated via the Multifunctional Ca2+-Calmodulin-Dependent Protein Kinase in Human Epithelial Cells," Journal of Physiology, 488:37-55 (1995).
Braun et al., "The Multifunctional Calcium/Calmodulin-Dependent Protein Kinase: From Form to Function," Annual Review of Physiology, 57:417-445 (1995).
De Ferrari et al, "Distinct Patterns of Calcium Transients During Early and Delayed Afterdepolarizations Induced by Isoproterenol in Ventricular Myocytes," Circ., 91:2510-2515 (1995).
De Koninck et al., "Sensitivity of CaM Kinase II to the Frequency of Ca2+ Oscillations," Science, 279:227-230 (1998).
Ellenbogen et al. "Safety and Efficacy of Intravenous Diltiazem in Atrial Fibrillation or Atrial Flutter", The American Journal of Cardiology 75:45-49 (Jan. 1, 1995).
Enslen et al., "Characterization of Ca2+/Calmodulin-Dependent Protein Kinase IV," The Journal of Biological Chemistry, 269:15520-15527 (1994).
File Caplus on STN, DN No. 121:277477. Okakazi et al. "KN-62, a specific Ca+ /calmodlulin-dependent protien kinase inhibitor, reversibly depresses teh rate of beating of cultured fetal mouse cardiac myocytes' J. Pharmacol. Exp. Thr. vol. 270, No. 3, pp., 1994.* *
File CAplus on STN, No. 109:142429. Meerson et al. "Prevention of Ischemic and Reperfusion Arrhythmias with Calmodulin Blocker, Trifluperazine,' Kardiologiya. vol. 28, No. 5, pp. 91-95, 1988.* *
File CAplus on STN, No. 114:159690. Yasugawa et al. "Autophosphorylation of Calcium/Calmodulin Dependent Protein Kinase II: Effects on Interaction Between Enzyme and Substrate', Jpn. J. Pharmacology, vol. 55, No. 2, pp. 263-274. abstract only, 1991.* *
File CAplus on STN, No. 122:314463. Hidaka et al. JP 06293730. Abstract only, 1995.* *
File Medline on STN. AN No. 77049877. Matsubara et al. "Experimental Anti Arrhythmic Effects of a New Beta Adrnergic Receptor Agent, dl-I-(tert. butylamino)-3-[(2-propinyloxy)phenoxy]2-propanol hydrochloride (dl Ko 1400-Cl). " Nippon Yakurigaku Zasshi. Folia pharmacologica Japonica (Jul. 1976) vol. 72, No. 5, pp. 557-571. Abstract only.* *
File Toxlit on STN. DN No. CA-114-156919Q. Cheng et al. "Effects of a New Intracellular Calcium Antagonist, KT-362, on Delayed Afterdepolarization oand Triggered Activity in Guinea Pig Ventricular Myocardium" Yakuri to Chiryo. vol. 18, Supp. 13, p. s5313-s3514. Abstract Only. (1990).* *
Goldenberg et al. "Intravenous Diltiazem for the Treatment of Patients with Atrial Fibrillation or Flutter and Moderate to Severe Congestive Heart Failure" The American Journal of Cardiology 74:884-889 (Nov. 1, 1994).
Griffith et al., "Inhibition of Calcium/Calmodulin-Dependent Protein Kinase in Drosophila Disrupts Behavioral Plasticity," Neuron, 10:501-509 (1993).
Ishida et al. "Inactivation of Ca2+/Calmodulin-Dependent Protein Kinase II by Ca 2+/Calmodulin" Journal of Biochemistry 115(6):1075-1082 (1994).
Ishikawa et al. "Effect of a New Ca2+-Calmodulin-Dependent Protein Kinase II Inhibitor on GABA Release in Cerebrospinal Fluid of the Rat" The Journal of Pharmacology and Experimental Therapeutics 254(2):598-602 (1990).
Ishikawa et al. "Effect of New CA+ 2-Calmodulin-Dependent Protein Kinase II Inhibitor on Gaba Release in Cerebrospinal Fluid of the Rat', J. of Pharm. and Exp. Ther. vol. 254, No. 2, pp. 598-602, 1990.* *
Lee et al., "Effect of Ischemia on Calcium-Dependent Fluorescence in Rabbit Hearts Containing Indo 1," Circulation, 78:1047-1059 (1988).
Levin et al. "Mechanism by Which Psychotropic Drugs Inhibit Adenosine Cyclic 3',5'-Monophosphate Phosphodiesterase of Brain" Molecular Pharmacology 12:581-589 (1976).
Levin et al. "Mechanism by Which Psychotropic Drugs Inhibit Adenosine Cyclic 3′,5′-Monophosphate Phosphodiesterase of Brain" Molecular Pharmacology 12:581-589 (1976).
Li et al., "The Effect of Ca2+-Calmodulin-Dependent Protein Kinase II on Cardiac Excitation-Contraction Coupling in Ferret Ventricular Myocytes," Journal of Physiology, 501:17-31 (1997).
Okazaki et al. "KN-62, a Specific Ca++/ Calmodulin-Dependent Protein Kinase Inhibitor, Reversibly Depresses the Rate of Beating of Cultured Fetal Mouse Cardiac Myocytes " The Journal of Pharmacology and Experimental Therapeutics 270(3):1319-1324 (1994).
Prozialeck et al. "Photoaffinity Labeling of Calmodulin by Phenothiazine Antipsychotics" Molecular Pharmacology 19:264-269 (1981).
Rich et al. "Substrate-directed Function of Calmodulin in Autophosphorylation of Ca2+/Calmodulin-dependent Protein Kinase II" The Journal of Biological Chemistry 273(43):28424-28429 (Oct. 23, 1998).
Roufogalis "Phenothiazine Antagonism of Calmodulin: A Structurally-Nonspecific Interaction" Biochemical and Biophysical Research Communications 98(3):607-613 (Feb. 12, 1981).
Schaffer et al. "Phenothiazine Protection in Calcium Overload-Induced Heart Failure: A Possible Role For Calmodulin" The American Physiological Society H328-H334 (1983).
Sumi et al., "The Newly Synthesized Selective Ca2+/Calmodulin Dependent Protein Kinase II Inhibitor KN-93 Reduces Dopamine Contents in PC12h Cells," Biochem. Biophys. Res. Comm., 181:968-975 (1991).
Thandroyen et al., "Intracellular Calcium Transients and Arrhythmia in Isolated Heart Cells," Circ. Res., 69:810-819 (1991).
Tieleman et al., "Verapamil Reduces Tachydcardia-induced Electrical Remodeling of the Atria," Circulation, 95:1945-1953 (1997).
Tokumitsu et al. "KN-62, 1-[N,O-Bis(5-isoquinolinesulfonyl)-N-methyl-L-tyrosyl]-4-phenylpiperazine, a Specific Inhibitor of Ca2+/Calmodulin-dependent Protein Kinase II" The Journal of Biological Chemistry 265(8):4315-4320 (Mar. 15, 1990).
Waxman et al. "Verapamil for Control of Ventricular Rate in Paroxysmal Supraventricular Tachycardia and Atrial Fibrillation or Flutter" Annals of Internal Medicine 94(1):1-6 (1/81).
Weiss et al. "Interaction of Calmodulin with Psychotropic Drugs: Neuropsychopharmacological Implications" Psychopharmacology Bulletin 19(3)378-386 (1983).
Weiss et al. "Interaction of Drugs with Calmodulin: Biochemical, Pharmacological and Clinical Implications" Biochemical Pharmacology 31(13):2217-2226 (1982).
Weiss et al. "Mechanism for Selectively Inhibiting the Activation of Cyclic Nucleotide Phosphodiesterase and Adenylate Cyclase by Antipsychotic Agents" Advances in Cyclic Nucleotide Research 9:285-303 (1978).

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040073262A1 (en) * 2000-11-02 2004-04-15 Cardiac Pacemakers, Inc. Technique for discriminating between coordinated and uncoordinated cardiac rhythms
US20040138648A1 (en) * 2000-12-18 2004-07-15 Cardiac Pacemakers, Inc. Drug delivery system for implantable medical device
US20030069606A1 (en) * 2001-06-15 2003-04-10 Girouard Steven D. Pulmonary vein stent for treating atrial fibrillation
US7209783B2 (en) 2001-06-15 2007-04-24 Cardiac Pacemakers, Inc. Ablation stent for treating atrial fibrillation
US20030018362A1 (en) * 2001-06-15 2003-01-23 Chris Fellows Ablation stent for treating atrial fibrillation
US20030060854A1 (en) * 2001-09-25 2003-03-27 Qingsheng Zhu Evoked response sensing for ischemia detection
US20040266675A1 (en) * 2001-10-01 2004-12-30 Mark Anderson Use of calmodulin kinase II inhibitors to treat myocardial dysfunction in structural heart disease
AU2002341938B2 (en) * 2001-10-01 2007-08-30 Vanderbilt University Use of calmodulin kinase II inhibitors to treat myocardial dysfunction in structural heart disease
US7320959B2 (en) * 2001-10-01 2008-01-22 Vanderbilt University Use of calmodulin kinase II inhibitors to treat myocardial dysfunction in structural heart disease
US20030153951A1 (en) * 2002-02-08 2003-08-14 Ideker Raymond E. Methods and devices for treating arrhythmias using defibrillation shocks
US20030216349A1 (en) * 2002-04-18 2003-11-20 Luiz Belardinelli Method for treating arrhythmias
US7005425B2 (en) * 2002-04-18 2006-02-28 Cv Therapeutics, Inc. Method for treating arrhythmias
US8275456B2 (en) 2002-11-12 2012-09-25 Cardiac Pacemakers, Inc. Implantable device for delivering cardiac drug therapy
US20040093034A1 (en) * 2002-11-12 2004-05-13 Girouard Steven D. Implantable device for delivering cardiac drug therapy
US20040158290A1 (en) * 2002-11-30 2004-08-12 Girouard Steven D. Method and apparatus for cell and electrical therapy of living tissue
US20040158289A1 (en) * 2002-11-30 2004-08-12 Girouard Steven D. Method and apparatus for cell and electrical therapy of living tissue
US20040220203A1 (en) * 2003-02-10 2004-11-04 Zaijie Wang Method and composition for potentiating an opiate analgesic
US7256200B2 (en) * 2003-02-10 2007-08-14 The Board Of Trustees Of The University Of Illinois, A Body Corporate And Politic Of The State Of Illinois Method and composition for potentiating an oplate analgesic
US20050043675A1 (en) * 2003-08-21 2005-02-24 Pastore Joseph M. Method and apparatus for modulating cellular metabolism during post-ischemia or heart failure
US8016783B2 (en) 2003-08-21 2011-09-13 Cardiac Pacemakers, Inc. Method and apparatus for modulating cellular metabolism during post-ischemia or heart failure
US20050137626A1 (en) * 2003-12-19 2005-06-23 Pastore Joseph M. Drug delivery system and method employing external drug delivery device in conjunction with computer network
US20100113563A1 (en) * 2004-01-30 2010-05-06 Zaijie Wang Method for Treating Pain with a Calmodulin Inhibitor
US7840263B2 (en) 2004-02-27 2010-11-23 Cardiac Pacemakers, Inc. Method and apparatus for device controlled gene expression
US20050192637A1 (en) * 2004-02-27 2005-09-01 Girouard Steven D. Method and apparatus for device controlled gene expression
US20050245972A1 (en) * 2004-04-29 2005-11-03 Medtronic, Inc. Apparatus and methods for treating arrhythmia and lowering defibrillation threshold
US7764995B2 (en) 2004-06-07 2010-07-27 Cardiac Pacemakers, Inc. Method and apparatus to modulate cellular regeneration post myocardial infarct
US20060079445A1 (en) * 2004-06-16 2006-04-13 Pitt Geoffrey S CaMKII/calcium channel binding-related compositions and methods
WO2006007444A2 (en) * 2004-06-16 2006-01-19 The Trustees Of Columbia University In The City Of New York CaMKII/CALCIUM CHANNEL BINDING-RELATED COMPOSITIONS AND METHODS
WO2006007444A3 (en) * 2004-06-16 2006-12-14 Univ Columbia CaMKII/CALCIUM CHANNEL BINDING-RELATED COMPOSITIONS AND METHODS
US20060057639A1 (en) * 2004-06-16 2006-03-16 Pitt Geoffrey S CaMKII/calcium channel phosphorylation-related compositions and methods
US7828711B2 (en) 2004-08-16 2010-11-09 Cardiac Pacemakers, Inc. Method and apparatus for modulating cellular growth and regeneration using ventricular assist device
US20060036126A1 (en) * 2004-08-16 2006-02-16 Jeffrey Ross Method and apparatus for modulating cellular growth and regeneration using ventricular assist device
US8805492B2 (en) 2004-08-20 2014-08-12 Cardiac Pacemakers, Inc. Method and apparatus for delivering combined electrical and drug therapies
US20090259268A1 (en) * 2004-08-20 2009-10-15 Gregory Waimong Chan Method and apparatus for delivering combined electrical and drug therapies
US7981065B2 (en) 2004-12-20 2011-07-19 Cardiac Pacemakers, Inc. Lead electrode incorporating extracellular matrix
US20060136028A1 (en) * 2004-12-20 2006-06-22 Jeffrey Ross Epicardial patch including isolated extracellular matrix with pacing electrodes
US8060219B2 (en) 2004-12-20 2011-11-15 Cardiac Pacemakers, Inc. Epicardial patch including isolated extracellular matrix with pacing electrodes
US20090149900A1 (en) * 2005-04-11 2009-06-11 Julia Moffitt Transvascular neural stimulation device
US20110082537A1 (en) * 2005-04-11 2011-04-07 Julia Moffitt Transvascular neural stimulation device
US8929990B2 (en) 2005-04-11 2015-01-06 Cardiac Pacemakers, Inc. Transvascular neural stimulation device and method for treating hypertension
US7979141B2 (en) 2005-05-16 2011-07-12 Cardiac Pacemakers, Inc. Transvascular reshaping lead system
US20100016927A1 (en) * 2005-05-16 2010-01-21 Anthony Caparso Transvascular reshaping lead system
US8126561B2 (en) 2005-10-24 2012-02-28 Cardiac Pacemakers, Inc. Implantable and rechargeable neural stimulator
US8634921B2 (en) 2005-10-24 2014-01-21 Cardiac Pacemakers, Inc. Implantable and rechargeable neural stimulator
US8660648B2 (en) 2005-10-24 2014-02-25 Cardiac Pacemakers, Inc. Implantable and rechargeable neural stimulator
US20070093875A1 (en) * 2005-10-24 2007-04-26 Cardiac Pacemakers, Inc. Implantable and rechargeable neural stimulator
US7616990B2 (en) 2005-10-24 2009-11-10 Cardiac Pacemakers, Inc. Implantable and rechargeable neural stimulator
US9951061B2 (en) 2013-03-06 2018-04-24 Allosteros Therapeutics, Inc. CaMKII inhibitors and uses thereof
US10759792B2 (en) 2014-09-05 2020-09-01 The Johns Hopkins University CaMKII inhibitors and uses thereof
US11325908B2 (en) 2014-09-05 2022-05-10 The Johns Hopkins University CaMKII inhibitors and uses thereof

Also Published As

Publication number Publication date
AU6257798A (en) 1998-08-25
ATE218854T1 (de) 2002-06-15
ES2178157T3 (es) 2002-12-16
EP0966274B1 (en) 2002-06-12
DE69805973T2 (de) 2002-12-19
US7251525B2 (en) 2007-07-31
EP0966274A2 (en) 1999-12-29
WO1998033491A3 (en) 1999-02-18
US20030134777A1 (en) 2003-07-17
DE69805973D1 (de) 2002-07-18
WO1998033491A2 (en) 1998-08-06
JP2001509808A (ja) 2001-07-24

Similar Documents

Publication Publication Date Title
US6518245B1 (en) Treatment of arrhythmias via inhibition of a multifunctional calcium/calmodulin-dependent protein kinase
Doba et al. Role of central and peripheral adrenergic mechanisms in neurogenic hypertension produced by brainstem lesions in rat
CA2201784C (en) Method and formulation of stimulating nitric oxide synthesis
TWI508726B (zh) 治療心房纖維性顫動之方法
Gauthier et al. Acute and chronic cardiovascular effects of 6-hydroxydopamine in dogs
Patterson et al. Ventricular fibrillation in a conscious canine preparation of sudden coronary death--prevention by short-and long-term amiodarone administration.
Hirakata et al. Angiotensins and the failing heart. Enhanced positive inotropic response to angiotensin I in cardiomyopathic hamster heart in the presence of captopril.
JP2004500906A (ja) 電気的調節および化学的調節のための心臓デバイス、ならびに心肺障害の処置のための血管作用性腸ペプチド
US7781402B2 (en) Methods and implantable devices for treating supraventricular arrhythmias
US4271192A (en) Process for treatment and prevention of ventricular fibrillation
Bonnardeaux et al. Action of angiotensin and analogues on the heart
Wilber et al. Postinfarction sudden death: significance of inducible ventricular tachycardia and infarct size in a conscious canine model
CA2051369C (en) Thyroid hormone cardiac treatment
Jones et al. Effects of lidocaine and verapamil on defibrillation in humans
Miyazaki et al. Modulation of cardiac autonomic neurotransmission by epicardial superfusion. Effects of hexamethonium and tetrodotoxin.
US5512592A (en) Method of producing cardiotonic effect and improving cardiac contractile function by administration of carnosine
Malfatto et al. The response to overdrive pacing of triggered atrial and ventricular arrhythmias in the canine heart.
Gilbert et al. Influence of sympathomimetic pressor drugs on arrhythmias caused by multiple stimuli
Caffrey Enkephalin inhibits vagal control of heart rate, contractile force and coronary blood flow in the canine heart in vivo
US5840762A (en) Method for the treatment of cardiac arrhythmias and shortening of action potential duration
Barlow et al. Vagotonic effects of enkephalin are not mediated by sympatholytic mechanisms
Morcos et al. Direct myocardial effects of cocaine
DEGUCHI et al. Ganglion stimulating action of candicine
Siden et al. Epicardial propranolol administration for ventricular arrhythmias in dogs: matrix formulation and characterization
EP1261346A1 (en) Modulation of cardiovascular injury

Legal Events

Date Code Title Description
FPAY Fee payment

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: PAT HOLDER CLAIMS SMALL ENTITY STATUS, ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: LTOS); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FPAY Fee payment

Year of fee payment: 8

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20150211